You are on page 1of 11

MOLECULAR AND CELLULAR BIOLOGY, Dec. 1995, p. 7106–7116 Vol. 15, No.

12
0270-7306/95/$04.0010
Copyright q 1995, American Society for Microbiology

Degradation of the Proto-Oncogene Product c-Fos by the Ubiquitin


Proteolytic System In Vivo and In Vitro: Identification and
Characterization of the Conjugating Enzymes
ILANA STANCOVSKI,1 HEDVA GONEN,1 AMIR ORIAN,1 ALAN L. SCHWARTZ,2
1
AND AARON CIECHANOVER *

Department of Biochemistry and Rappaport Institute for Research in the Medical Sciences, Faculty of Medicine,
Technion-Israel Institute of Technology, Haifa 31096, Israel,1 and Division of Hematology-Oncology,
Children’s Hospital, and Washington University School of Medicine, St. Louis, Missouri 631102
Received 26 July 1995/Returned for modification 12 September 1995/Accepted 15 September 1995

The transcription factor c-Fos is a short-lived cellular protein. The levels of the protein fluctuate signifi-
cantly and abruptly during changing pathophysiological conditions. Thus, it is clear that degradation of the
protein plays an important role in its tightly regulated activity. We examined the involvement of the ubiquitin
pathway in c-Fos breakdown. Using a mutant cell line, ts20, that harbors a thermolabile ubiquitin-activating
enzyme, E1, we demonstrate that impaired function of the ubiquitin system stabilizes c-Fos in vivo. In vitro, we
reconstituted a cell-free system and demonstrated that the protein is multiply ubiquitinated. The adducts serve
as essential intermediates for degradation by the 26S proteasome. We show that both conjugation and
degradation are significantly stimulated by c-Jun, with which c-Fos forms the active heterodimeric transcrip-
tional activator AP-1. Analysis of the enzymatic cascade involved in the conjugation process reveals that the
ubiquitin-carrier protein E2-F1 and its human homolog UbcH5, which target the tumor suppressor p53 for
degradation, are also involved in c-Fos recognition. The E2 enzyme acts along with a novel species of
ubiquitin-protein ligase, E3. This enzyme is distinct from other known E3s, including E3a/UBR1, E3b, and
E6-AP. We have purified the novel enzyme ;350-fold and demonstrated that it is a homodimer with an
apparent molecular mass of ;280 kDa. It contains a sulfhydryl group that is essential for its activity,
presumably for anchoring activated ubiquitin as an intermediate thioester prior to its transfer to the substrate.
Taken together, our in vivo and in vitro studies strongly suggest that c-Fos is degraded in the cell by the
ubiquitin-proteasome proteolytic pathway in a process that requires a novel recognition enzyme.

The protein products of c-fos and c-jun function coopera- bers of the family. The viral counterparts of c-fos and c-jun
tively as components of the mammalian transcription factor (v-fos and v-jun, respectively) have been identified as onco-
AP-1 (reviewed in reference 1). The proteins form a stable genes in transforming retroviruses (10, 37). In addition, dereg-
heterodimeric complex mediated via a leucine zipper associa- ulated expression of both c-Fos and c-Jun leads to neoplastic
tion that binds to a palindromic DNA sequence known as the transformation of rat fibroblasts (12, 54). It appears therefore
12-O-tetradecanoylphorbol 13-acetate (TPA)-responsive ele- that the stability of Fos determines to a large extent the activity
ment or the AP-1 binding site. Transcription of both c-fos and of AP-1 and may be important for the proper control of cell
c-jun is induced by TPA and other activators of protein kinase proliferation.
C (1), resulting in increased biosynthesis of the two proteins. A series of previous observations suggest that at least two
Regulation of transcription and posttranslational modifica- proteolytic pathways may be involved in c-Fos breakdown. (i)
tions, such as phosphorylation, control the activity of AP-1 in Using a cell-free system, Hirai and colleagues (27) and Carillo
response to extracellular stimuli. The composition of the AP-1 and colleagues (5) demonstrated the involvement of calpains
complex changes from predominantly Jun homodimers prior in the breakdown of c-Fos and c-Jun. (ii) In a reticulocyte
to induction to mostly Jun-Fos heterodimers following induc- lysate, c-Fos is slightly degraded in an ATP-dependent manner
tion. Fos proteins (c-Fos, FosB, Fra-1, and Fra-2) are thought (5, 8, 45). After interaction with phosphorylated c-Jun, c-Fos is
to modulate AP-1 activity, as they cannot form homodimers rapidly degraded by a trans degradation signal (45), but the
but only heterodimers with members of the Jun family proteins identity of the proteolytic system involved has remained ob-
(c-Jun, JunB, and JunD). The heterodimer has higher DNA- scure. In the case of Jun-independent degradation, it has been
binding and transcriptional activity than the Jun homodimer
suggested that the process is mediated by the ubiquitin-pro-
(11). Inhibition of the activity of members of the Fos family,
teasome system: immunodepletion of the ubiquitin-activating
either by expression of antisense transcript or following micro-
enzyme, E1, inhibited degradation of c-Fos, whereas addition
injection of specific antibodies, blocked the induction of cell
of purified enzyme restored activity (8). However, even in this
proliferation and cell cycle progression (29, 47). However,
gene disruption experiments (34, 63) suggest that the absence case, multiply ubiquitinated species of c-Fos, the essential in-
of the c-fos gene is probably compensated for by other mem- termediates in the process, have not been demonstrated, and
the enzymes involved in conjugation and degradation have not
been identified. Treier and colleagues suggested a role for the
* Corresponding author. Mailing address: Department of Biochem- ubiquitin pathway in regulating the stability of c-Jun (59) and
istry, Faculty of Medicine, Technion-Israel Institute of Technology, demonstrated that targeting is mediated via the d domain in
P.O. Box 9649, Haifa 31096, Israel. Phone: 972-4-295365, 972-4- the c-Jun molecule. Interestingly, v-Jun, which is a stable pro-
295379, or 972-4-295356. Fax: 972-4-513922 or 972-4-552296. tein, lacks the d domain and escapes ubiquitination and sub-

7106
VOL. 15, 1995 c-Fos DEGRADATION BY THE UBIQUITIN SYSTEM 7107

sequent degradation. The researchers proposed that this sta- terial expression plasmid encoding the UbcH5 (50) was obtained from Martin
bilization plays a role in the oncogenicity of v-Jun. However, Scheffner (Deutsches Krebsforschungszentrum, Heidelberg, Germany).
Degradation of c-Fos protein in vivo. Exponentially growing cells were seeded
Jariel-Encontre and colleagues have recently shown that ubi- at 5 3 105 cells per 65-mm dish. After 16 h, the medium was changed to DMEM
quitination is not required for degradation of the c-Jun pro- containing 0.5% FCS, and incubation was continued for additional 48 h. Induc-
tein, which can be degraded by the 26S proteasome without tion of c-Fos was accomplished by the addition of DMEM containing 20% FCS
prior ubiquitination (31). for 1 h. For labeling, cells were washed with methionine-free medium and
The ubiquitin-dependent pathway plays an important role in incubated at 308C for 15 min in the same medium containing 20% dialyzed FCS
and 300 mCi of [35S]methionine per ml. Labeling of the cells was continued for
the degradation of short-lived regulatory proteins, such as cy- additional 20 min at either 30 or 43.58C (pulse). Cultures were harvested (0 min)
clins (reviewed in reference 40), the tumor suppressor protein or incubation was continued (following removal of the label; chase) at 30 or
p53 (9, 49, 50–53), and the MATa2 repressor (28). In addition, 39.58C for 30 and 60 min in a complete medium containing 4 mM unlabeled
the system is also involved in limited proteolysis, such as in the methionine. Cells were washed twice in ice-cold phosphate-buffered saline (PBS)
and lysed in 1 ml of ice-cold RIPA buffer (PBS containing 1% Nonidet P-40,
maturation of p105, the precursor of the NF-kB transcriptional 0.5% sodium deoxycholate, 0.1% sodium dodecyl sulfate [SDS], 2 mM phenyl-
activator (43), and probably in processing of antigens restricted methylsulfonyl fluoride [PMSF], and 5 mg of aprotinin per ml). Following cen-
to class I major histocompatibility complex (MHC) molecules trifugation at 13,000 3 g for 30 min, aliquots from the supernatant that contained
(38). Degradation of a protein via the ubiquitin system involves equal amounts of protein were immunoprecipitated with anti-Fos antibody
SC-52 (Santa Cruz Biotechnology), resolved by SDS-polyacrylamide gel electro-
several discrete steps. In this process, ubiquitin is first activated phoresis (PAGE), and analyzed by fluorography.
by the ubiquitin-activating enzyme E1. The activated molecule Fractionation of crude reticulocyte lysate, preparation of E1 and E2-F1 en-
is transferred to one of several ubiquitin carrier proteins (E2s, zymes, and initial characterization of the Fos conjugating ligase E3. Reticulo-
or ubiquitin-conjugating enzymes [UBCs]), which transfers it cyte-rich blood was induced in rabbits by successive injections of phenylhy-
drazine, and reticulocyte lysate was prepared as described before (25). Lysates
to a third enzyme, ubiquitin-protein ligase, E3. This enzyme were resolved by anion-exchange chromatography over DEAE-cellulose (What-
catalyzes the last step in the process, covalent conjugation of man) into unadsorbed material (fraction I) and high-salt eluate (fraction II) as
ubiquitin to the protein substrate that bound to the ligase (7, described before (25). E1 was purified to homogeneity from fraction II following
52). Following formation of a multiubiquitin chain, the tagged affinity chromatography over immobilized ubiquitin (25). E2-F1 was purified to
homogeneity from fraction I as described before (2). Recombinant UbcH5, the
protein is degraded by the 26S proteasome complex, with the human homolog of E2-F1 (50) (cDNA cloned into pET3a vector) was purified
release of free and reutilizable ubiquitin (reviewed in refer- from bacterial extracts (following induction) as follows. Extract (from 1 liter of
ences 7, 14, and 32). cultured cells) was chromatographed on DEAE anion-exchange resin. Fraction
Using a cell line harboring a thermolabile E1 enzyme, we I (;75 mg of protein) was concentrated (to ;2.5 ml) with a Centriprep 10
(Amicon) and resolved on a HiLoad Superdex 75 HR (16 by 600 mm) with a fast
demonstrate that c-Fos is stabilized at the restrictive temper- protein liquid chromatography (FPLC) system (Pharmacia) in a buffer contain-
ature. Dissection of the proteolytic pathway in a cell-free sys- ing 20 mM Tris-HCl (pH 7.2), 2 mM dithiothreitol (DTT), and 150 mM NaCl
tem reveals that c-Fos, which is a short-lived protein in the cell, (buffer A). Fractions (2.4 ml) were collected, and the protein was visualized by
is multiply ubiquitinated and that the high-molecular weight Coomassie brilliant blue staining after SDS-PAGE of aliquots from the various
fractions. Salt was removed by repeated concentration-dilution cycles with Cen-
conjugates serve as intermediates for proteolysis by the 26S tricon-10 microconcentrators (Amicon) and buffer A without NaCl. The UbcH5-
proteasome complex. We also demonstrate that the process is containing fractions were concentrated to ;100 ml and pooled. We obtained ;1
mediated by a recently described ubiquitin carrier protein, mg of .85% homogeneous protein. For initial characterization of the E3 en-
E2-F1 (2), and a novel, as yet unidentified, species of E3. The zyme, rabbit reticulocyte fraction II was precipitated by (NH4)2SO4 into fractions
IIA (0 to 38%) and IIB (42 to 80%). Fraction IIA (2 ml, 20 mg/ml) was loaded
E2-F1 enzyme can be functionally replaced by a recombinant onto a Superdex 200 HR gel filtration chromatography column (16 by 600 mm)
human homolog, UbcH5 (50). Both conjugation and degrada- and resolved and treated as described for UbcH5. Conjugation of c-Fos was
tion are strongly stimulated by c-Jun. Taken together, the data determined in the presence of 2.5-ml aliquots as described below.
strongly suggest that the ubiquitin pathway is involved in reg- Purification of the E3 protein. (i) Anion-exchange chromatography. Fraction
IIA (200 mg) was loaded onto a Resource Q (Pharmacia) anion-exchange col-
ulating the stability of the transcription factor c-Fos, both in umn (16 by 120 mm). The column was equilibrated with 20 mM Tris-HCl (pH
vivo and in vitro, and thus plays an important role in regulating 7.2), 180 mM KCl, and 1 mM DTT. After loading, the column was washed
the cellular level of AP-1 complex and its transcriptional ac- thoroughly with the equilibrating buffer. Adsorbed proteins were eluted with a
tivity. 500-ml linear salt gradient to 500 mM KCl. Fractions (20 ml) were collected, and
salt was removed with Centriprep-30 concentrators as described above. Fos-
conjugating activity (E3-Fos) was eluted at ;240 to 280 mM KCl.
(ii) Gel filtration chromatography. Pooled and concentrated fractions from
MATERIALS AND METHODS the first purification step were loaded onto a Superdex 200 HR gel filtration
column (16 by 600 mm), and proteins were resolved as described above. Frac-
Cell lines. The Chinese hamster cell line ts20, a temperature-sensitive cell tions containing E3-Fos activity (proteins eluted at ;280 kDa) were pooled and
cycle-arrested mutant, and its parental cell line E36 were previously described dialyzed against a buffer containing 20 mM Tris-HCl (pH 7.2), 1 M (NH4)2SO4,
(36). ts20 cells harbor a thermolabile ubiquitin-activating enzyme, E1, and ubi- and 1 mM DTT (buffer B).
quitin conjugation is significantly decreased at the nonpermissive temperature. (iii) Hydrophobic chromatography. Pooled and dialyzed fractions from the
Consequently, degradation of short-lived proteins in these cells is also impaired second purification step were loaded onto a Bio-Rad Econo-Pac Methyl-HIC
(19). The ts20.E1.C2 (ts-E1) cell line (a generous gift from Patricia M. Handley- hydrophobic column (5-ml bed volume) equilibrated in buffer B. Unadsorbed
Gearhart, Washington University School of Medicine [WUMS], St. Louis, Mo.) proteins were washed with buffer B. A 100-ml linear gradient [1.0 to 0.0 M
was obtained by transfection of ts20 cells with a human wild-type E1 cDNA (21). (NH4)2SO4] was used to elute bound proteins. Salt was removed and fractions
The B-cell lymphoma line C3.F6 (62) was obtained from Osami Kanagawa, were concentrated as described above. E3-Fos activity was eluted at ;500 mM
WUMS. Cells were grown as a monolayer at 308C (E36, ts20, and ts-E1) or in salt.
suspension at 378C (C3.F6) in Dulbecco’s modified Eagle’s medium (DMEM) (iv) Hydroxylapatite chromatography. A portion of the hydrophobic-chroma-
supplemented with 10% fetal calf serum (FCS). Transfections were carried out tography-eluted E3 was dialyzed against a buffer containing 10 mM KPi (pH 7.4)
by electroporation (64). The permissive and nonpermissive temperatures used and 1 mM DTT and loaded onto a Bio-Rad Econo-Pac HTP (hydroxylapatite;
for the experiments with the various E1 cell lines were 30 and 39.58C, respec- 5-ml bed volume) column equilibrated in the same buffer. Adsorbed proteins
tively. were eluted with a 100-ml linear gradient to 600 mM KPi. E3-Fos activity was
Plasmids. The cDNAs for rat c-fos in pSP65 and human papillomavirus type eluted at ;320 to 410 mM KPi.
16 E6 in pGEM-2 vectors have been described elsewhere (8, 9). The v-fos- (v) Anion-exchange chromatography. The remaining portion of the hydropho-
containing plasmid was constructed by replacement of the NcoI-AvrII fragment bic-chromatography-eluted E3 was dialyzed against a buffer containing 20 mM
from the c-fos cDNA in the pSP65 vector with the respective fragment from the Tris-HCl (pH 7.2), 200 mM KCl, and 1 mM DTT and loaded onto a Mono Q
FBJ-2 vector (13). In vitro transcription-translation reactions for generation of (Pharmacia) anion-exchange chromatography column (1-ml bed volume) equil-
[35S]methionine-labeled proteins were performed in a TNT-coupled wheat germ ibrated in the same buffer. E3-Fos protein was eluted at ;300 to 340 mM KCl
extract system (Promega) according to the manufacturer’s instructions. The with a 200 to 600 mM linear gradient.
pKH6 vector (44, 45) containing a hexahistidine (6-His)-tagged human c-jun was The protein concentration was determined throughout the purification by the
a generous gift from Dirk Bohmann (EMBL, Heidelberg, Germany). The bac- Bradford method (3) with bovine serum albumin as a standard. Protein compo-
7108 STANCOVSKI ET AL. MOL. CELL. BIOL.

sition was determined by SDS-PAGE and Coomassie brilliant blue or silver


staining. c-Fos conjugation was determined as described below.
Conjugation and degradation of [35S]methionine-labeled proteins. [35S]me-
thionine-labeled substrates were generated in vitro in a coupled transcription-
translation reaction in wheat germ extract (TNT; Promega). Conjugation and
degradation assays were performed essentially as described before (9). Briefly,
the reaction mixture contained, in a final volume of 25 ml, crude reticulocyte
lysate (10 ml; ;1 mg of protein) or fraction II (;100 mg of protein) and purified
E2-F1 (0.2 mg), 40 mM Tris-HCl (pH 7.6), 5 mM MgCl2, 2 mM DTT, 5 mg of
ubiquitin, and [35S]methionine-labeled substrate (;30,000 cpm). In the conju-
gation assays, 1 mg of ubiquitin aldehyde (26) and 5 mM adenosine 59-O-(3-
thiotriphosphate) (ATP-g-S) (Boehringer, Mannheim, Germany) were added.
Reaction mixtures without ATP contained 0.5 mg of hexokinase and 10 mM
2-deoxyglucose (9). For degradation, ATP (0.5 mM) and the ATP-regenerating
system (10 mM phosphocreatine and 5 mg of creatine kinase) were added.
Conjugation reactions were incubated for 30 min at 378C, and degradation
reactions were carried out at the same temperature for 2 h. Following incubation,
reaction mixtures were resolved via SDS-PAGE, and the gels were fluoro-
graphed, dried, and exposed to BioMax X-Ray film (Kodak) or to a phosphor-
imaging plate scanner (Fuji, Tokyo, Japan) for quantitative evaluation. The
radioactivity incorporated into adducts was expressed as a percentage of total
radioactivity.
Degradation of c-Fos conjugates by the 26S proteasome. For analysis of the
role of the 26S proteasome complex in the degradation of c-Fos-ubiquitin con-
jugates, adducts were synthesized as described above in a 100-ml reaction mix-
ture. Following incubation, enzymes were inactivated by the addition of NaOH
to a final concentration of 0.1 N and further incubation for 10 min. The pH was
neutralized by the addition of HCl, and the conjugates were dialyzed at 48C
against a buffer containing 20 mM Tris-HCl (pH 7.2) and 2 mM DTT. Degra- FIG. 1. Metabolic stability of c-Fos in E1-defective cell line. Endogenous
dation of c-Fos conjugates was assayed in the presence of purified 26S protea- c-Fos protein was induced by 20% FCS, and cells were pulse-labeled with
some complex as described before (18, 56). Briefly, reaction mixtures contained, 35
[ S]methionine for 35 min as described in Materials and Methods. Inactivation
in a final volume of 50 ml, conjugates (;10,000 cpm), 40 mM Tris-HCl (pH 7.6), of E1 was carried out by incubating the cells at 43.58C for 20 min. Cell lysates
5 mM MgCl2, and 2 mM DTT. The 26S proteasome complex (1.5 mg) and 1 mM were prepared without further incubation (0 min) or after incubation at 30 or
ATP were added as indicated. Following incubation at 378C for 2 h, conjugate 39.58C for 30 and 60 min (chase). Immunoprecipitates with anti-Fos antibody
degradation was determined by quantifying the remaining adducts with a phos- were analyzed by SDS-PAGE and fluorography.
phorimaging plate scanner. In parallel, release of radioactivity into the trichlo-
roacetic acid-soluble fraction was also monitored. Since the preparation of the
conjugates also contains free, unreacted labeled c-Fos, degradation of an equal
amount of radiolabeled c-Fos was monitored in parallel.
Formation of 125I-ubiquitin thioester with E1, E2-F1, and E3-Fos. Ubiqui- E36 at both temperatures (;40 min; quantitative analysis not
tin-E1 and ubiquitin-E2 thioester adducts were generated and detected with
125
shown). Inhibition of E1 resulted in three- to fourfold prolon-
I-ubiquitin as described before (2). To obtain the E3-Fos-ubiquitin thioester, gation of the half-life of the protein (analysis not shown). The
E3 (5 mg) was added to a reaction mixture that also contained E1, E2-F1,
125
I-ubiquitin, and ATP. partial stabilization is due, most probably, to the leakiness of
Purification of recombinant His-Jun. The 6-His-c-Jun protein was purified the mutation and to the inability to inhibit completely the E1
from transfected HeLa or E36 cell lines with a nickel chelate column (Ni21- enzyme. This problem has previously been discussed in detail
nitrilotriacetic acid-agarose; Quiagen) as previously described (44, 45). (19) (see also below). Incubation of the cells at the nonper-
Preparation of methylated ubiquitin. Reductively methylated ubiquitin
(MeUb) was prepared from bovine ubiquitin (Sigma) by a previously described missive temperature did not affect viability (.95%) (19). To
procedure (24). Fluorescamine determination of free amino groups revealed that ensure that c-Fos stabilization was not due to a change in other
more than 95% of the moieties were blocked. factors or parameters in the mutant cell line, we used an
Preparation of cell extracts. Cells were washed twice in ice-cold PBS and E1-rescued cell line, ts-E1. This cell line was obtained by stable
incubated on ice for 15 min in a hypotonic lysis buffer (20 mM sodium-HEPES
[N-2-hydroxyethylpiperazine-N9-2-ethanesulfonic acid, pH 7.5], 10 mM KCl, 1.5 correction of the defect in the E1 enzyme following transfec-
mM MgCl2, 2 mM DTT; 2 ml for 5 3 108 cells). Lysis was completed with a tion with an expression vector carrying wild-type human E1
Dounce homogenizer and controlled under the microscope. Nuclei were re- cDNA (21). As depicted in Fig. 1, c-Fos was rapidly degraded
moved and cytosol was prepared as described before (15). Extracts contained ;5 in the ts-E1 cell line at the nonpermissive temperature, cor-
to 10 mg of protein per ml.
roborating the involvement of E1 in destabilizing the protein.
We noted the appearance of high-molecular-mass c-Fos-
RESULTS related proteins in the wild-type, mutant, and wild-type E1-
transfected cells at the low temperature and in the wild-type
Functional ubiquitin-activating enzyme E1 is essential for and transfected cells at the high temperature. These bands are
c-Fos degradation in vivo. To evaluate the potential role of the most probably ubiquitin conjugates of c-Fos, as they disappear
ubiquitin system in the turnover of the c-Fos transcription in the mutant cells at the high temperature. We were able to
factor in vivo, we used ts20 cells, a cell cycle-arrested mutant detect a slight increase in these labeled proteins following the
derived from the E36 cell line that harbors a thermolabile addition of the proteasome inhibitor MG115 (not shown; for
ubiquitin-activating enzyme, E1 (36). At the nonpermissive the inhibitor, see below). However, because of their low abun-
temperature, conjugation of ubiquitin is significantly dimin- dance, we could not detect them directly with antiubiquitin
ished. Consequently, the degradation of the bulk of short-lived antibody and Western immunoblot analysis. It is possible that,
proteins is also inhibited (19). We monitored the degradation unlike adducts of other proteins, such as those of the cyclin-
of c-Fos at both the permissive temperature (308C) and after a dependent kinase inhibitor p27 (42), the conjugates of c-Fos
shift to the restrictive temperature (39.58C). As shown in Fig. are more sensitive to isopeptidases and thus do not accumulate
1, the c-Fos protein was stable in the mutant cell line, ts20, at to a detectable level even when the proteasome is inhibited.
the restrictive temperature but was rapidly degraded at the Thus, at this point we can speculate on their identity but not
permissive temperature. Quantitative measurement of c-Fos prove it unambiguously.
radioactivity indicated that the half-life of the protein in ts20 It should be noted that stabilization of any single short-lived
cells at 308C is similar to that measured in the parental cell line protein in the E1 mutant cells does not unequivocally demon-
VOL. 15, 1995 c-Fos DEGRADATION BY THE UBIQUITIN SYSTEM 7109

degradation by the 26S proteasome complex (35). Ubiquitin


aldehyde inhibits certain isopeptidases (26). Under these con-
ditions, the steady-state level of ubiquitin adducts increases,
facilitating their detection. Next, we tested the formation of
c-Fos conjugates in a fractionated lysate. As shown in Fig. 2B
(lane 1), incubation of c-Fos in the presence of fraction II does
not lead to formation of conjugates. Fraction II (the high-salt
eluate obtained during anion-exchange chromatography of cell
extracts) contains most of the conjugating enzymes and the 26S
protease. However, ubiquitin and certain ubiquitin-conjugat-
ing enzymes, E2s, are contained in fraction I, the material that
does not adsorb to the resin. Addition of ubiquitin to fraction
II had only a slight effect on the formation of conjugates (Fig.
2B, lane 2). Addition of homogeneously purified E2-F1 led to
a significant increase in conjugate formation (Fig. 2B, lane 3).
E2-F1 is a ubiquitin-conjugating enzyme involved in targeting
non-‘‘N-end rule’’ substrates (2), including the tumor suppres-
sor protein p53 (9). To extend these results to a more relevant
physiological system, extracts from C3.F6, a B-cell lymphoma
cell line, were fractionated in a similar manner. As can be seen
FIG. 2. E2-F1- and UbcH5-dependent conjugation of ubiquitin to c-Fos. in Fig. 2C, the formation of c-Fos conjugates is similar to that
Conjugation of ubiquitin to [35S]methionine-labeled c-Fos was monitored by observed in the reticulocyte lysate. E2-F1 is a rabbit enzyme,
SDS-PAGE and fluorography as described in Materials and Methods. (A) Ubiq- homologous to several evolutionarily conserved members of
uitin conjugation to c-Fos in a crude reticulocyte lysate. Incubation was carried the E2 family. It is similar to Saccharomyces cerevisiae UBC4
out in the absence (lane 1) or presence (lane 2) of ATP-g-S. (B) Ubiquitin
conjugation to c-Fos in fraction II. Lane 1, fraction II; lane 2, fraction II plus and UBC5 (55), Drosophila melanogaster UbcD1 (58), Triticum
ubiquitin; lane 3, fraction II, ubiquitin, and purified E2-F1. (C) Ubiquitin con- aestivum UBC8 (17), and several enzymes from Arabidopsis
jugation to c-Fos in lymphocyte cell extract. Cytosolic cell extract was prepared thaliana (16). It has been shown that the wheat germ enzyme
as described in Materials and Methods and resolved on DEAE-cellulose. The accounts for most of the conjugating activity in extracts derived
reaction mixtures in the different lanes are the same as those in panel B. (D)
c-Fos conjugation in the presence of recombinant UbcH5. c-Fos was incubated
from the plant (17). Recently, UbcH5, a human member of this
in the presence of reticulocyte fraction II and ubiquitin only (lane 1) or plus subfamily, was characterized and cloned (50). The enzyme is
purified E2-F1 (lane 2) or 0.5 mg of recombinant UbcH5 (lane 3). Ori, origin of also involved in targeting of p53 (50). Bacterially expressed
the gel; conj, conjugates. UbcH5 could functionally replace E2-F1 protein in promoting
c-Fos conjugation (Fig. 2D). Since UbcH5, UBC8, and E2-F1
can also catalyze E6-dependent conjugation of p53, it is pos-
strate direct involvement of the ubiquitin pathway in its pro- sible that members of this highly conserved E2 family have a
teolysis. One can always argue that stabilization of the target similar function in the cell and play an important role in the
protein is due to an upstream effect that results from E1 inac- regulation of stability of many cellular proteins.
tivation. However, such an effect is not known and cannot be Influence of MeUb on c-Fos conjugation. To further evalu-
analyzed at our current state of knowledge. The in vivo effect ate the role of ubiquitin in c-Fos conjugation, we used a de-
must therefore be supported by additional experimental evi- rivative of the protein in which the amino groups had been
dence derived from the establishment of a faithful cell-free blocked by reductive methylation, methylated ubiquitin
system. (MeUb). It was previously shown that MeUb is efficiently li-
Ubiquitination of c-Fos in vitro. Degradation of a protein gated to protein substrates but cannot form polyubiquitin
via the ubiquitin system is a complex process that involves chains (24). Thus, MeUb can be used as a chain terminator in
specific enzymatic components and primary proteolysis recog- the process of ubiquitin conjugation. The effect of different
nition signals on the target substrate. In addition, targeting can concentrations of MeUb on c-Fos conjugation was examined in
also involve ancillary proteins and secondary, posttranslational the experiment shown in Fig. 3. [35S]methionine-labeled c-Fos
modifications that render the substrate susceptible to recogni- was subjected to conjugation in a partially purified in vitro
tion. Thus, the initial observation that E1 is involved in c-Fos system containing E1, E2-F1, and fraction IIA as a source of
degradation in vitro (8) and in vivo (see above) still leaves E3 in the presence of 5 mg of ubiquitin. As can be seen in Fig.
numerous important unresolved problems. The ubiquitin con- 3, increasing concentrations of MeUb significantly inhibited
jugates which are the essential intermediates in the process c-Fos conjugate formation. From quantitative evaluation of the
have not been demonstrated. The conjugating enzymes have results presented in Fig. 3, the concentration of MeUb re-
not been identified, and their mechanism of action has not quired for 50% inhibition was threefold higher than the cor-
been dissected. The proteolysis recognition signals of the sub- responding concentration of ubiquitin, as was described previ-
strate are still obscure, and the potential role of c-Jun in tar- ously (23).
geting c-Fos for conjugation has not been studied. Thus, it was Effect of c-Jun on ubiquitin-dependent degradation and con-
important to establish an in vitro system that will reproduce jugation of c-Fos. It has been shown previously that c-Fos
faithfully the ubiquitin-mediated proteolytic process. As can be degradation is targeted by a phosphorylation-dependent signal
seen in Fig. 2, c-Fos can generate high-molecular-mass cova- on c-Jun (45). The researchers proposed a model in which a
lent conjugates with ubiquitin. In the experiment presented in signal-induced dephosphorylation of c-Jun (for example, fol-
Fig. 2A, the c-Fos substrate was incubated in a reticulocyte lowing treatment of cells with TPA) transiently stabilizes c-
lysate in the absence (lane 1) and presence (lane 2) of ATP- Fos–c-Jun dimers in stimulated cells. After withdrawal of the
g-S. To enhance the formation of conjugates, the reaction stimulating agent and phosphorylation of c-Jun, AP-1 activity
contained ATP-g-S (instead of ATP) and ubiquitin aldehyde. decreases as a result of protein degradation (45). However, the
ATP-g-S promotes conjugation by E1 but does not support system that mediates the process and the mechanism(s)
7110 STANCOVSKI ET AL. MOL. CELL. BIOL.

FIG. 3. Inhibition of ubiquitin conjugation to c-Fos by MeUb. [35S]methi-


onine-labeled c-Fos was subjected to ubiquitin conjugation in a reticulocyte
lysate as described in Materials and Methods. MeUb was added at the indicated
concentrations, and the reaction mixtures were resolved via SDS-PAGE. The
chart represents quantitative analysis of the data by phosphorimaging plate
scanner analysis. FIG. 4. Effect of c-Jun on degradation and conjugation of c-Fos. (A) c-Fos
degradation. Degradation of [35S]methionine-labeled c-Fos was assayed in a
reticulocyte lysate as described in Materials and Methods. Control reactions
without ATP were incubated at either 08C (lane 1) or 378C (lane 2). Lanes 3 to
6, reaction mixtures containing ATP were incubated at 378C in the absence (lane
through which c-Jun targets c-Fos for degradation have not 3) or presence (lanes 4 to 6) of the indicated amounts of His-tagged c-Jun. (B)
been identified. We investigated the possibility that c-Jun is c-Fos conjugation. Conjugation of labeled c-Fos was carried out essentially as
involved in the conjugation of ubiquitin to c-Fos, leading to described in Materials and Methods in the presence of a partially purified system
containing E1, E2-F1, and fraction IIA in the absence (lane 1) or presence of
subsequent degradation of the protein. trans recognition has ATP (lanes 2 to 4) and the indicated amounts of His-Jun (lanes 3 and 4). Ori,
been previously described in the case of the tumor suppressor origin of the gel; conj, conjugates.
p53: the protein is targeted for ubiquitin conjugation and deg-
radation by the human papillomavirus E6 oncoprotein (50).
We therefore purified 6-His-tagged c-Jun (His-Jun) (44, 45) tor. However, it was not clear that the modification leads to the
and assessed its effect on in vitro degradation of c-Fos. As degradation of the protein and that the protease involved is
demonstrated in Fig. 4A, and in agreement with previous data indeed the 26S proteasome complex. Previous studies have
(45), increasing concentrations of purified His-Jun target la- shown that a peptide-aldehyde, MG115 (carbobenzoxyl-leuci-
beled c-Fos for degradation. In order to identify the step in the nyl-leucinyl-norvalinal-H), is a potent and rather specific in-
degradation pathway that is affected by c-Jun, we analyzed the hibitor of the 20S proteasome (43, 48). In vitro-translated
effect of purified His-Jun on the conjugation of ubiquitin to [35S]methionine-labeled c-Fos was incubated in cytosolic cell
c-Fos. The reaction was carried out in a partially purified extracts at 378C for 2 h (Fig. 5). Potential calpain-dependent
system containing labeled c-Fos, ubiquitin-activating enzyme proteolysis was inhibited by treatment of the extract with the
E1, ubiquitin carrier protein E2-F1, and fraction IIA as a calcium chelator EGTA (ethylene glycol tetraacetic acid) at 3
source of ubiquitin-protein ligase E3. It is clear that addition of mM (5) (MgCl2 was added to the reaction mixture at a con-
His-Jun increases the formation of high-molecular-weight c- centration of 10 mM). Addition of MG115 markedly inhibited
Fos conjugates (Fig. 4B). It should be noted that ubiquitin is c-Fos degradation (Fig. 5, lanes 4 and 5). It should be noted
conjugated to the labeled substrate even in the absence of that in the presence of ATP (Fig. 5, lanes 3 to 5), a higher-
His-Jun (Fig. 4B, lane 2). However, the efficiency of degrada- molecular-mass form of c-Fos accumulated. This change is due
tion of c-Fos is significantly lower in the absence of c-Jun. It is
possible that the detection of the c-Jun effect on conjugate
formation is more difficult than monitoring its effect on deg-
radation because the adducts are also metabolized by isopep-
tidases not inhibited by ubiquitin aldehyde. Alternatively, it is
also possible that Jun affects other, as yet unidentified, ubiqui-
tin-independent proteolytic pathway(s) (see above). Stabiliza-
tion of c-Fos in E1-inactivated cells makes the possibility that
such putative pathways are operating in the cell rather unlikely.
The 26S proteasome is required for c-Fos degradation. The FIG. 5. Inhibition of the 26S proteasome blocks c-Fos degradation. Degra-
dation of [35S]methionine-labeled c-Fos was carried out in a lymphocyte extract
results described above demonstrate that the c-Fos protein for 2 h as described in Materials and Methods. Lanes 1 and 2, incubations at 0
forms conjugates with ubiquitin, suggesting a role for ubiq- and 378C, respectively, in the absence of ATP. Lanes 3 to 5, incubations at 378C
uitin-mediated proteolysis of this short-lived transcription fac- in the presence of ATP. MG115 was added at the indicated concentrations.
VOL. 15, 1995 c-Fos DEGRADATION BY THE UBIQUITIN SYSTEM 7111

FIG. 6. Identification of the E3 enzyme involved in c-Fos conjugation. (A) c-Fos conjugation. Fraction IIA was resolved via gel filtration chromatography over a
Superdex 200 HR column as described in Materials and Methods. E3 activity was monitored in the presence of E1, E2-F1, ubiquitin, ATP-g-S, and ubiquitin aldehyde,
in the presence of aliquots derived from the resolved fractions. (B) p53 conjugation. Conjugation of p53 was monitored as described in the legend to panel A in the
presence of in vitro-translated, unlabeled E6. Lane A, [35S]methionine-labeled substrates incubated in the presence of purified E1 and E2-F1. Lane B, control reactions
incubated as described for lane A but with the addition of 10 mg of unresolved fraction IIA. Other lanes, reaction mixtures incubated in the presence of E1, E2-F1,
and aliquots from the resolved fractions indicated above the lanes. The positions of molecular size markers and labeled substrates are marked. Arrows represent
fractions with the highest activity. Ori, origin of the gel; conj, conjugates.

to a posttranslational modification, most probably phosphory- resin at neutral pH and was purified from the unadsorbed
lation, that can significantly alter the migration of c-Fos in material (fraction I). The enzyme is not specific to c-Fos and is
SDS-PAGE (8, 12). The high-molecular-mass bands could be involved in the degradation of a variety of substrates, p53, for
precipitated by an anti-c-Fos antibody following incubation of example (9, 50, 51). In a previous study, Scheffner and col-
in vitro-translated unlabeled protein in the presence of leagues demonstrated that the E2 enzyme transfers ubiquitin
[32P]ATP (not shown). from E1 to a cysteine moiety in the E3 molecule E6-AP (E6-
To investigate whether c-Fos-ubiquitin adducts are essential associated protein), involved in conjugation of p53 (52). The
intermediates in the proteolytic process and to further corrob- activated ubiquitin residue is then transferred to the substrate
orate the role of the 26S proteasome complex in their degra- that is specifically bound to E3 (7, 52). For initial character-
dation, we generated high-molecular-mass conjugates of labeled ization of the E3 protein involved in recognition of the c-Fos
c-Fos. The enzymes used for conjugation were inactivated by transcription factor, we first tested whether the wheat germ
the addition of NaOH followed by neutralization with HCl. extract used for in vitro translation of the labeled substrate
The conjugates were then subjected to proteolysis in the pres- contains the ligase. Ubiquitin conjugates could not be detected
ence of purified 26S proteasome complex (56). c-Fos conju- upon incubation of the substrate alone (not shown) or follow-
gates were degraded in an ATP-dependent manner by purified ing addition of purified E1 and E2-F1 (Fig. 6A, lane A). In-
26S proteasome complex. The unmodified protein was not creasing the concentration of the wheat germ extract yielded a
recognized by the 26S proteasome (data not shown). The per- similar negative result. We concluded that the E3 enzyme
cent degradation was 0% without 26S or ATP, 4% 6 0.2% with required for c-Fos conjugation was not present in the wheat
26S and without ATP, and 48% 6 2.7% with 26S and ATP germ extract. It should be noted that E6-AP, the E3 enzyme
(assayed in duplicate; range shown). involved in p53 conjugation, is also absent from the wheat
Identification of the ubiquitin-protein ligase E3, involved in germ extract (51). We observed that the E3 protein necessary
c-Fos conjugation. Molecular analysis of the complex mecha- for ubiquitination of c-Fos is present in fraction IIA (Fig. 6A,
nisms of degradation of c-Fos requires purification and char- lane B; data for other reticulocyte lysate subfractions not
acterization of the enzymes involved. We have shown (Fig. 2) shown).
that the recently described ubiquitin carrier enzyme E2-F1 (2) To further characterize the ligase involved, we resolved re-
is essential for c-Fos conjugation. Unlike most members of the ticulocyte lysate fraction IIA by gel filtration chromatography
E2 family, this protein did not adsorb to the anion-exchange over a Superdex 200 column. Samples from the concentrated
7112 STANCOVSKI ET AL. MOL. CELL. BIOL.

were assayed for E3 activity based on their ability to conjugate


ubiquitin to [35S]methionine-labeled c-Fos in the presence of
purified E1 and E2-F1 enzymes. In parallel, the protein profile
of the different fractions isolated during purification was mon-
itored by SDS-PAGE and Coomassie brilliant blue staining.
The peak of activity eluted from all columns comigrates (in
SDS-PAGE) with a protein of ;140 kDa. Figure 8 presents the
peak of activity recovered from the last two purification steps
along with the profiles of the proteins resolved by electro-
phoresis. It is clear that the indicated 140-kDa protein eluted
from the different resins comigrates with the activity. It was
important to follow the activity and protein profile in all col-
umns, as the E3 enzyme has not been purified to homogeneity.
Taken together, the results strongly suggest that the 140-kDa
protein is indeed E3-Fos (see also below). A summary of the
purification procedure is given in Table 1: 0.1 mg of protein
was isolated, with an overall purification of ;335-fold and a
recovery of ;2.0%. An attempt to sequence the protein from
the N-terminal residue failed, suggesting that the molecule is
blocked at this residue. Eight peptides derived from tryptic
hydrolysis of the E3-Fos enzyme were subjected to Edman
degradation, and their sequences revealed no significant ho-
mology with any known protein (not shown). Cloning of the
gene and expression of the protein will be essential for un-
equivocal identification of the enzyme as the c-Fos ligase and
for a detailed analysis of the mechanism of action of this novel
E3 protein.
Characterization of E3-Fos ubiquitin-protein ligase. Ubiqui-
tination of c-Fos involves the concerted action of the ubiqui-
tin-activating enzyme E1, the ubiquitin-conjugating enzyme
FIG. 7. Scheme for purification of E3-Fos. Reticulocyte lysate was applied to
a DEAE-cellulose resin and further resolved as described in Materials and
E2-F1, and a novel ubiquitin-protein ligase, E3. Using gel
Methods. filtration chromatography, we have demonstrated that the E3
protein has an apparent molecular mass of ;280 kDa. It is
probably a homodimer, as the molecular mass of the SDS-
PAGE-resolved protein is ;140 kDa. The polypeptide chains
fractions were analyzed for formation of [35S]methionine-la- of the dimer are not linked via -S-S- bonds, as electrophoresis
beled c-Fos conjugates in the presence of purified E1 and of the protein in a denaturing environment but in the absence
E2-F1 enzymes (Fig. 6A). In parallel, we examined the same of b-mercaptoethanol revealed the same 140-kDa monomer.
fractions for conjugation of [35S]methionine-labeled p53 in the We investigated the possibility that E3-Fos, like E6-AP (52),
presence of unlabeled, in vitro-translated E6 (Fig. 6B). As contains an active sulfhydryl (SH) group involved in ubiquitin
depicted in Fig. 6A, c-Fos-conjugating activity peaked in frac- binding and transfer. As can be seen in Fig. 9, the activity of the
tions 25 and 26, corresponding to an apparent molecular mass enzyme was inhibited significantly by the alkylating agent N-
of ;260 to 280 kDa. In striking contrast, p53-conjugating ac- ethylmaleimide. To demonstrate that the 140-kDa protein is
tivity peaked at fraction 28, corresponding to a molecular mass indeed the E3 enzyme and to further corroborate the role of
of ;120 kDa. This activity represents, most probably, E6-AP the SH group in mediating ubiquitin transfer, we generated a
(30). In addition, a truncated, bacterially expressed form of thioester of the enzyme with labeled ubiquitin in the presence
E6-AP that is active in p53 conjugation (a generous gift from of E1, E2, and ATP (2, 20). As shown in Fig. 10, incubation of
Martin Scheffner) (50) was inactive in c-Fos conjugation (data E3-Fos in the presence of the two other enzymes leads to the
not shown). Purified E3a (46) was also inactive in the presence formation of a thiol group-sensitive adduct between ubiquitin
of both 14-kDa E2 (the N-end rule E2, the mammalian ho- and the ligase. This product had a molecular mass of ;160 kDa
molog of the yeast UBC2/RAD6 protein [33]) and E2-F1 (not (Fig. 10, lane 5). Addition of excess free thiol groups (b-
shown; see also below). The c-Fos-conjugating ligase is also mercaptoethanol) results in the disappearance of most of the
different from the N-end rule E3b (22) because it does not radioactivity (Fig. 10, lane 6). The remaining label migrated at
interact with 14-kDa E2 (not shown). Taken together, these ;150 kDa. This SH-insensitive compound is probably an
findings clearly indicate that the enzyme that conjugates c-Fos autoubiquitination product of the enzyme. This hypothesis can
is a novel protein distinct from the several known ligases in- only be tested following cloning and expression of the enzyme.
volved in conjugation of p53 and N-end rule substrates. Not surprisingly, the E1-ubiquitin thioester was also labile to
Purification of the ubiquitin ligase E3-Fos. The initial iden- the addition of the reducing agent (Fig. 10, lanes 1 and 3). We
tification of a novel E3 activity involved in c-Fos conjugation to were able to detect the E2-ubiquitin adduct (lane 2) only fol-
ubiquitin (designated E3-Fos) prompted further purification lowing a shorter exposure. It is ‘‘hidden’’ in the gel shown by
and characterization of the protein. Reticulocyte lysate was the labeled ubiquitin.
used as a source for the enzyme. The purification scheme (Fig. It was important to further examine the specificity of recog-
7) involved a combination of anion-exchange (DEAE-cellu- nition of the novel E3 enzyme towards different substrates. The
lose, Pharmacia Resource, and Mono Q), gel filtration (Super- initial identification of the E3-Fos protein indicated that the
dex 200 HR), hydrophobic (Bio-Rad Methyl-HIC), and hydrox- activity is distinct from E6-AP, the ligase required for conju-
ylapatite (Bio-Rad HTP) chromatographies. Column fractions gation of p53, and from the N-end rule E3s, E3a and E3b (see
VOL. 15, 1995 c-Fos DEGRADATION BY THE UBIQUITIN SYSTEM 7113

FIG. 8. Comigration of c-Fos-conjugating activity with the putative 140-kDa E3-Fos. E3-Fos activity in the various fractions was determined by monitoring ubiquitin
conjugation to [35S]methionine-labeled c-Fos in the presence of E1 and E2-F1 enzymes as described in Materials and Methods. Conjugation was quantitated by
phosphorimaging plate scanner analysis and expressed as a percentage of total radioactivity incorporated into high-molecular-weight adducts. In parallel, samples from
the same fractions were resolved by SDS–7.5% PAGE, and the proteins were visualized by staining the gel with Coomassie brilliant blue. (A) Conjugation activity of
E3-Fos resolved on a hydrophobic (Methyl-HIC) resin. Peak fractions from the Superdex 200 gel filtration chromatography column were processed and resolved on
a Methyl-HIC column as described in Materials and Methods. Aliquots (2.5 ml) from the concentrated fractions were monitored for c-Fos conjugation. (B)
Electrophoretic analysis of E3-Fos-containing fractions resolved on the Methyl-HIC column. Aliquots (10 ml) were electrophoresed on a 7.5% polyacrylamide gel,
followed by staining with Coomassie brilliant blue. Lane M, molecular size markers. Lanes 11 to 19 represent the corresponding gradient fractions as described in
Materials and Methods. (C) Conjugation activity of E3-Fos resolved on hydroxylapatite (HTP). The activity was determined as described in the legend to panel A. (D)
Electrophoretic analysis of E3-Fos-containing fractions resolved on the HTP column. Aliquots from the corresponding fractions were processed as described in the
legend to panel B. The gel was stained with silver.

above). Accordingly, E3-Fos could not promote the conjuga- amino acids at the C terminus of the c-Fos protein. In partic-
tion of lysozyme and oxidized RNase A (E3a substrates) or of ular, the modification affected serine residues necessary for
protein S (E3b substrate) (not shown). Also, E3a could not phosphorylation. As a result, the extent of phosphorylation of
substitute for E3-Fos in c-Fos conjugation (see above). Con- v-Fos is markedly decreased. We tested the ability of v-Fos to
jugation of other substrates, such as histone H2A, actin, tro- be conjugated to ubiquitin. As can be seen in Fig. 11, conju-
ponin T, and MyoD, could not be stimulated by this ligase gation of v-Fos is much less efficient than that of c-Fos (about
either. These substrates are conjugated by a distinct and novel threefold) both in a crude lymphocyte extract (Fig. 11A) and in
E3 enzyme (17a). Interestingly, E3-Fos is involved in ubiquiti- a reconstituted system containing the purified conjugating en-
nation of c-Jun, the other component of the AP-1 transcription zymes (Fig. 11B). Not surprisingly, v-Fos is also stable in the
factor complex (not shown). This finding raises the possibility cell-free system (45), and ;80% of the protein can be detected
that both components of the complex are targeted by the same following 3 h of incubation (not shown).
machinery.
Conjugation recognition signal(s) in c-Fos. It was previously DISCUSSION
demonstrated that whereas the t1/2 of c-Fos in vivo is ;30 min,
the v-fos gene product is more stable and has a half-life of ;2 Many regulatory cellular proteins are extremely unstable. In
h (12). Interestingly, c-Jun does not affect v-Fos stability (45). the case of signal-inducible transcription factors, it is apparent
Nucleotide sequence analysis revealed that the v-fos gene dif- that changes in their steady-state levels can influence the ac-
fers from c-fos in that it has undergone an out-of-frame dele- tivity of the corresponding target genes. Thus, it is important to
tion of 104 bases (60, 61). Consequently, the v-Fos protein has understand the molecular mechanisms that govern the cellular
49 amino acids at its C terminus that are unrelated to the 48 stability of these proteins.
7114 STANCOVSKI ET AL. MOL. CELL. BIOL.

TABLE 1. Summary of the purification of E3-Fos ligasea


Total Total Sp act Purification Recovery
Fraction
protein (mg) activity (U) (U/mg) (fold) (%)

Fraction IIA 1,540 56,980 37 1 100


Resource Q 76 34,200 450 12.2 60
Superdex 200 25 16,175 647 17.5 28.3
Methyl-HIC 3.25 3,120 960 25.9 5.4
HTP 0.1 1,240 12,400 335.1 2.1
a
Two liters of reticulocyte lysate was used for purification. The protein con-
centration was determined by the Bradford method (3). Activity was determined
by measuring conjugation of [35S]methionine-labeled c-Fos in the presence of
E1, E2-F1, and the tested fractions as described in Materials and Methods.
Quantitation was performed with a phosphorimaging plate scanner and is ex-
pressed in units of activity. One unit represents incorporation of 1% of radio- FIG. 10. Formation of a thioester adduct between ubiquitin and purified
activity into conjugates per milligram of protein (determined in the linear range E3-Fos. The reactions were performed as described previously (2) and in Ma-
of the reaction). The resins used for the purification are: Resource Q, anion- terials and Methods. The samples contained E1 and 125I-ubiquitin alone (lanes
exchange resin (Pharmacia); Superdex 200 HR, gel filtration matrix (Pharmacia); 1 and 3) or with E2-F1 (lanes 2 and 4) or with E2-F1 and purified E3-Fos (lanes
Methyl-HIC, hydrophobic matrix (Bio-Rad); and HTP, hydroxylapatite beads 5 and 6). Reaction mixtures were incubated at 378C for 20 min and divided into
(Bio-Rad). two equal portions that were resolved via SDS-PAGE in the presence (1) or
absence (2) of b-mercaptoethanol (b-ME; 0.1 M), as indicated. The E1- and
E3-ubiquitin thioester adducts are indicated (E1-S;Ub and E3-S~Ub, respec-
tively), as is the unreacted radioactive ubiquitin (125I-Ub). E3F-Ub?, putative
In this study, we demonstrated that the degradation of the monoubiquitinated product of the E3-Fos ligase.
c-Fos transcription factor is mediated by the ubiquitin system
both in vivo and in vitro. Using a cell cycle-arrested mutant
that harbors a thermolabile ubiquitin-activating enzyme, E1, complement the in vivo findings, it was necessary to establish a
we show that c-Fos is stabilized at the nonpermissive temper- cell-free proteolytic system and study the different enzymes
ature. In contrast, the protein is unstable at the permissive and ancillary components involved.
temperature and at both temperatures in the wild-type cell. A We were able to reconstitute the c-Fos degradation pathway
mutant cell line that was rescued by transfection with a cDNA in vitro and to demonstrate that conjugation of ubiquitin to
encoding the wild-type E1 enzyme displays a wild-type pheno- c-Fos is an essential intermediate step in the proteolytic pro-
type. Using the same strategy, Chowdary and colleagues (6) cess. c-Fos is tagged by ubiquitin (Fig. 2), and the formation of
demonstrated the involvement of the ubiquitin system in the high-molecular-mass adducts is inhibited by MeUb (Fig. 3),
degradation of p53. However, it should be noted that in intact suggesting that targeting of the protein requires multiple ubi-
cells, it is difficult to determine whether the E1 enzyme is quitination. It is clear that these adducts serve as essential
directly involved in c-Fos conjugation and degradation or intermediates in the process, since only the conjugated protein
whether inactivation of the enzyme affects a different protein is recognized by the 26S proteasome and degraded (see above
that resides upstream in the proteolytic pathway of c-Fos. For and Fig. 5). Addition of c-Jun increases both conjugation and
example, E1 may be involved in the degradation of an uniden- subsequent degradation of c-Fos (Fig. 4). This is probably due
tified specific proteolytic inhibitor or an ancillary protein that to a physical interaction between the two proteins (45). It
stabilizes c-Fos: stabilization of the inhibitor or protein leads to should be noted that both conjugation and degradation pro-
stabilization of the target protein. Another alternative may ceed in the absence of c-Jun. However, the two processes,
involve targeting of c-Jun. The Jun protein, which is the other proteolysis in particular, are stimulated significantly following
component of the AP-1 transcription complex, is also targeted addition of the Jun protein. It is possible that the effect of
for degradation following ligation of ubiquitin (59). It is pos- c-Jun on conjugation is greater than the one that we detect and
sible that c-Jun is degraded first and thus exposes, in a second- that the conjugates formed are rapidly deubiquitinated by
ary manner, the c-Fos protein to the action of the ubiquitin
system or a ubiquitin-independent pathway. However, it was
recently reported that c-Jun can be efficiently degraded by the
26S proteasome without prior ubiquitination (31). To further
corroborate the role of ubiquitination in targeting c-Fos and to

FIG. 9. E3-Fos ubiquitin-ligase has an active SH group. Conjugation of ubiq-


uitin to c-Fos was monitored as described in Materials and Methods. The reac-
tion mixtures contained all three conjugating enzymes together with ATP-g-S.
For alkylation of SH groups, the indicated enzyme was incubated for 10 min at
228C in the presence of N-ethylmaleimide (NEM; 10 mM). The reagent was
neutralized by the addition of 6 mM DTT. Lane 1, complete reaction mixture; FIG. 11. Conjugation of ubiquitin to c-Fos and v-Fos proteins. Conjugation
lane 2, the E2-F1 enzyme pretreated with NEM and neutralized with DTT; lane reactions of the labeled substrates were performed as described in Materials and
3, E3-Fos pretreated with NEM and neutralized with DTT; lane 4, E3-Fos Methods. (A) Conjugation in crude lymphocyte extract. (B) Conjugation in a
pretreated with NEM that was previously incubated in the presence of DTT. Ori, reconstituted system containing E1, E2-F1, and E3-Fos (derived from the Mono
origin of the gel; conj, conjugates. Q purification step [Fig. 7]; 0.75 mg). Lanes 1, c-Fos; lanes 2, v-Fos.
VOL. 15, 1995 c-Fos DEGRADATION BY THE UBIQUITIN SYSTEM 7115

isopeptidases that are insensitive to ubiquitin aldehyde. The substrates for a Ca21-dependent calpain (5, 27). Our results, as
involvement of Jun represents yet another example of trans well as those of Treier and colleagues (59) studying the deg-
recognition and targeting. The tumor suppressor protein p53 is radation of c-Jun, indicate that the ubiquitin system is the main
targeted for degradation by the human papillomavirus onco- pathway that controls c-Fos and c-Jun stability.
protein E6 (53). Ornithine decarboxylase is targeted for deg- Control of stability of oncogene products and tumor sup-
radation by the 26S proteasome complex without prior ubiqui- pressors frequently contributes to transformation. Our results
tination in a process that requires antizyme (39). However, the provide additional evidence for the involvement of the ubiqui-
case of c-Fos–c-Jun dimer differs from the other known cases tin system in the breakdown of a key cellular regulator.
of trans-targeted degradation. In the p53-E6 complex, as well
as in the ornithine decarboxylase-antizyme complex, one sub- ACKNOWLEDGMENTS
unit of the binary complex provides the recognition site and
remains stable, whereas the other is targeted for degradation. We thank Dirk Bohmann (European Molecular Biology Laboratory,
In the c-Fos–c-Jun heterodimer, both components appear to Heidelberg, Germany) for providing the pKH6 plasmid; Julian Adams
be ubiquitinated. However, no evidence is available concerning and Ross Stein (MyoGenics, Inc., Cambridge, Mass.) for the protea-
some inhibitor MG115; Martin Scheffner (Deutsches Krebsforschungs-
the degradation of c-Jun adducts. Also, the Jun protein is an zentrum, Heidelberg, Germany) for the kind gift of the UbcH5 clone;
integral part of the AP-1 transcriptional complex and essential and Patricia M. Handley-Gearhart (Washington University School of
for its activity. This is not the case with E6 and antizyme. Medicine, St. Louis, Mo.) for the ts-E1 cells. We also thank Dganit
Therefore, it is expected that c-Jun is involved in regulating the Shkedy and Beatrice Bercovitch for help and advice. We are grateful
stability of the complex. to Daniel Schindler (The Weizmann Institute of Science, Rehovot,
Dissection of the enzymatic components involved in c-Fos Israel) for critically reading the manuscript.
conjugation and degradation reveals that conjugate synthesis is This research was supported by grants from the Council for Tobacco
catalyzed by the ubiquitin carrier protein E2-F1 and its human Research, Inc. (CTR), German-Israeli Foundation for Scientific Re-
homolog UbcH5 (Fig. 2). The 14-kDa E2, the N-end rule search and Development (G.I.F.), Israeli Academy of Humanities and
Sciences, Israel Cancer Research Fund (ICRF, USA), Israel Cancer
ubiquitin-activating enzyme, is significantly less active. This is Society, Foundation for Promotion of Research at the Technion, and
evident from the low conjugating activity of fraction II, which the Henri Gutwirth Fund for the Promotion of Research. I.S. was
contains the enzyme (Fig. 2B and C). Ubiquitination requires supported by a fellowship from the Lady Davis Foundation.
an additional novel protein that presumably functions as a
ubiquitin-protein ligase, E3 (Fig. 6). Purification of the novel REFERENCES
E3 activity revealed that the enzyme is a homodimer with an 1. Angel, P., and M. Karin. 1991. The role of Jun, Fos and the AP-1 complex
apparent molecular mass of ;280 kDa (Fig. 6 and 8). The in cell-proliferation and transformation. Biochim. Biophys. Acta 1072:129–
protein contains an active SH group that, like the E6-AP en- 157.
2. Blumenfeld, N., H. Gonen, A. Mayer, C. E. Smith, N. R. Siegel, A. L.
zyme, functions in the thioester cascade involving transfer of Schwartz, and A. Ciechanover. 1994. Purification and characterization of a
ubiquitin from E1 to E2-F1, from E2-F1 to E3-Fos, and then to novel species of ubiquitin-carrier protein, E2, that is involved in the degra-
the substrate (Fig. 9 and 10). E3-Fos is essential for conjuga- dation of a non-‘‘N-end rule’’ proteins. J. Biol. Chem. 269:9574–9581.
tion, suggesting involvement in recognition of the substrate. 3. Bradford, M. M. 1976. A rapid and sensitive method for quantitation of
microgram quantities of protein utilizing the principle of protein-dye bind-
The recognition appears to be specific, as the novel E3 does ing. Anal. Biochem. 72:248–254.
not conjugate the tumor suppressor protein p53 and a variety 4. Brown, K., S. Gerstberger, L. Carlson, G. Franzoso, and U. Siebenlist. 1995.
of N-end rule substrates and other proteins, such as actin, Control of IkB-a proteolysis by site-specific, signal-induced phosphorylation.
histone H2A, troponin T, and MyoD. We suspect, however, Science 267:1485–1488.
5. Carillo, S., M. Pariat, A.-M. Steff, P. Roux, M. Etienne-Julan, T. Lorca, and
that the enzyme is not entirely specific to c-Fos and that other M. Piechaczyk. 1994. Differential sensitivity of FOS and JUN family mem-
substrates will be discovered. We have already shown that it bers to calpains. Oncogene 9:1679–1689.
can conjugate c-Jun (not shown). An attractive model is that a 6. Chowdary, D. R., J. J. Dermody, K. K. Jha, and H. L. Ozer. 1994. Accumu-
single E2 enzyme interacts with a subset of E3s that recognize lation of p53 in a mutant cell line defective in the ubiquitin pathway. Mol.
Cell. Biol. 14:1997–2003.
common structural motifs on their protein targets. E2-F1 is 7. Ciechanover, A. 1994. The ubiquitin-proteasome proteolytic pathway. Cell
involved in the conjugation of both p53 (9, 50, 51) and c-Fos 79:13–21.
(this study), but the E3 enzymes involved are clearly distinct. 8. Ciechanover, A., J. A. DiGiuseppe, B. Bercovich, A. Orian, J. D. Richter,
The different E3 enzymes may not be specific to a single sub- A. L. Schwartz, and G. M. Brodeur. 1991. Degradation of nuclear oncopro-
teins by the ubiquitin system in vitro. Proc. Natl. Acad. Sci. USA 88:139–143.
strate, and it is likely that both E3-Fos and E6-AP recognize 9. Ciechanover, A., D. Shkedy, M. Oren, and B. Bercovich. 1994. Degradation
other proteins as well. As mentioned, our data suggest the of the tumor suppressor protein p53 by the ubiquitin-mediated proteolytic
involvement of E3-Fos in c-Jun conjugation. system requires a novel species of ubiquitin-carrier protein, E2. J. Biol.
The mode of recognition of c-Fos by this E3 enzyme is not Chem. 269:9582–9589.
10. Curran, T. 1988. The fos oncogene, p. 307–325. In E. P. Reddy, A. M. Skalka,
yet clear. In certain cases, specific phosphorylation or dephos- and T. Curran (ed.), The oncogene book. Elsevier, Amsterdam.
phorylation appears to target proteins for degradation. IkBa is 11. Curran, T., and B. R. Franza, Jr. 1988. Fos and Jun: the AP-1 connection.
targeted for degradation by the ubiquitin system following Cell 55:395–397.
signal transduction-induced phosphorylation of Ser-32 and 12. Curran, T., A. D. Miller, L. Zokas, and I. M. Verma. 1984. Viral and cellular
Fos proteins: a comparative analysis. Cell 36:259–268.
Ser-36 (4, 57). Mos is targeted for degradation by specific 13. Curran, T., G. Peters, C. van Bereven, N. M. Teich, and I. M. Verma. 1982.
dephosphorylation of Ser-3 (41). In the case of Fos, it has been FBJ murine osteosarcoma virus: identification and molecular cloning of
demonstrated that the product of the viral oncogene v-fos is biologically active proviral DNA. J. Virol. 44:674–682.
more stable than the cellular normal protein and that c-Jun 14. Finley, D., and V. Chau. 1991. Ubiquitination. Annu. Rev. Cell Biol. 7:25–69.
15. Flanagan, W. M., B. Corthésy, R. J. Bram, and G. R. Crabtree. 1991. Nuclear
does not affect its stability (45). The two proteins vary in the association of a T cell transcription factor blocked by FK-506 and cyclosporin
C-terminal domain, which contains multiple phosphorylation A. Nature (London) 352:803–807.
sites. We have shown that v-Fos is recognized less efficiently by 16. Girod, P.-A., T. P. Carpenter, S. van Nocker, M. L. Sullivan, and R. D.
the ubiquitin-conjugating machinery. Vierstra. 1993. Homologs of the essential ubiquitin-conjugating enzymes
UBC1, 4 and 5 in yeast are encoded by a multigene family in Arabidopsis
We could not rule out completely the partial involvement of thaliana. Plant J. 3:545–552.
another proteolytic system(s) in the breakdown of c-Fos. For 17. Girod, P.-A., and R. D. Vierstra. 1993. A major ubiquitin-conjugating system
example, it has been suggested that both c-Jun and c-Fos are in wheat germ extracts involves a 15-kDa ubiquitin-conjugating enzyme (E2)
7116 STANCOVSKI ET AL. MOL. CELL. BIOL.

homologous to the yeast UBC4/UBC5 gene products. J. Biol. Chem. 268: 42. Pagano, M., S. W. Tam, A. M. Theodoras, P. Beer-Romero, G. Del Sal, V.
955–960. Chau, P. Renée Yew, G. F. Draetta, and M. Rolfe. 1995. Role of the ubiqui-
17a.Gonen, H., and A. Ciechanover. Unpublished data. tin-proteasome pathway in regulating abundance of the cyclin-dependent
18. Gonen, H., A. L. Schwartz, and A. Ciechanover. 1991. Purification and kinase inhibitor p27. Science 269:682–685.
characterization of a novel protein that is required for degradation of N-a- 43. Palombella, V. J., O. J. Rando, A. L. Goldberg, and T. Maniatis. 1994. The
acetylated proteins by the ubiquitin system. J. Biol. Chem. 266:19221–19231. ubiquitin-proteasome pathway is required for processing of NF-kB1 precur-
19. Gropper, R., R. A. Brandt, S. Elias, C. F. Bearer, A. Mayer, A. L. Schwartz, sor protein and the activation of NF-kB. Cell 78:773–785.
and A. Ciechanover. 1991. The ubiquitin-activating enzyme, E1, is required 44. Papavassiliou, A. G., K. Bohmann, and D. Bohmann. 1992. Determining the
for stress-induced lysosomal degradation of cellular proteins. J. Biol. Chem. effect of inducible protein phosphorylation on the DNA-binding activity of
266:3602–3610. transcription factors. Anal. Biochem. 203:302–309.
20. Haas, A. L., J. V. Warms, A. Hershko, and I. A. Rose. 1982. Ubiquitin- 45. Papavassiliou, A. G., M. Treier, C. Chavrier, and D. Bohmann. 1992. Tar-
activating enzyme: mechanism and role in protein-ubiquitin conjugation. J. geted degradation of c-Fos, but not v-Fos, by a phosphorylated signal on
Biol. Chem. 257:2543–2548. c-Jun. Science 258:1941–1944.
21. Handley-Gearhart, P. M., J. S. Trausch-Azar, A. Ciechanover, and A. L. 46. Reiss, Y., and A. Hershko. 1990. Affinity purification of ubiquitin-protein
Schwartz. 1994. Rescue of the complex temperature-sensitive phenotype of ligase on immobilized protein substrates. J. Biol. Chem. 265:3685–3690.
Chinese hamster ovary E36ts20 cells by expression of the human ubiquitin- 47. Riabowol, K. T., R. J. Vosatka, E. B. Ziff, N. J. Lamb, and J. R. Feramisco.
activating enzyme cDNA. Biochem. J. 304:1015–1020. 1988. Microinjection of Fos-specific antibodies blocks DNA synthesis. Mol.
22. Heller, H., and A. Hershko. 1990. A ubiquitin-protein ligase specific for type Cell. Biol. 8:1670–1676.
III protein substrates. J. Biol. Chem. 265:6532–6535. 48. Rock, K. L., C. Gramm, L. Rothstein, K. Clark, R. Stein, L. Dick, D. Hwang,
23. Hershko, A., D. Ganoth, J. Pehrson, R. E. Palazzo, and L. H. Cohen. 1991. and A. L. Goldberg. 1994. Inhibitors of the proteasome block degradation of
Methylated ubiquitin inhibits cyclin degradation in clam embryo extracts. J. most cell proteins and the generation of peptides presented on MHC class I
Biol. Chem. 266:16376–16379. molecules. Cell 78:761–771.
24. Hershko, A., and H. Heller. 1985. Occurrence of a polyubiquitin structure in 49. Rolfe, M., P. Beer-Romero, S. Glass, J. Eckstein, I. Berdo, A. Theodoras, M.
ubiquitin-protein conjugates. Biochem. Biophys. Res. Commun. 128:1079– Pagano, and G. Draetta. 1995. Reconstitution of p53-ubiquitinylation reac-
1086. tions from purified components: the role of human ubiquitin-conjugating
25. Hershko, A., H. Heller, S. Elias, and A. Ciechanover. 1983. Components of enzyme UBC4 and E6-associated protein (E6-AP). Proc. Natl. Acad. Sci.
the ubiquitin-protein ligase system: resolution, affinity purification, and role USA 92:3264–3268.
in protein breakdown. J. Biol. Chem. 258:8206–8214. 50. Scheffner, M., J. M. Huibregtse, and P. M. Howley. 1994. Identification of a
26. Hershko, A., and I. A. Rose. 1987. Ubiquitin-aldehyde: a general inhibitor of human ubiquitin-conjugating enzyme that mediates the E6-AP-dependent
ubiquitin recycling processes. Proc. Natl. Acad. Sci. USA 84:1829–1833. ubiquitination of p53. Proc. Natl. Acad. Sci. USA 91:8797–8801.
27. Hirai, S., H. Kawasaki, M. Yaniv, and K. Suzuki. 1991. Degradation of 51. Scheffner, M., J. M. Huibregtse, R. D. Vierstra, and P. M. Howley. 1993. The
transcription factors, c-Jun and c-Fos, by calpain. FEBS Lett. 287:57–61. HPV-16 E6 and E6-AP complex function as a ubiquitin-protein ligase in the
28. Hochstrasser, M., M. J. Ellison, V. Chau, and A. Varshavsky. 1991. The ubiquitination of p53. Cell 75:495–505.
short-lived MATa2 transcriptional regulator is ubiquitinated in vivo. Proc. 52. Scheffner, M., U. Nuber, and J. M. Huibregtse. 1995. Protein ubiquitination
Natl. Acad. Sci. USA 88:4606–4610. involving an E1-E2-E3 enzyme ubiquitin thioesther cascade. Nature (Lon-
29. Holt, J. C., T. Venkat Gopal, A. D. Moulton, and A. W. Nienhuis. 1986. don) 373:81–83.
Inducible production of c-Fos antisense RNA inhibits 3T3 cell proliferation. 53. Scheffner, M., B. A. Werness, J. M. Huibregtse, A. J. Levine, and P. M.
Proc. Natl. Acad. Sci. USA 83:4794–4798. Howley. 1990. The E6 oncoprotein encoded by human papillomavirus types
30. Huibregtse, J. M., M. Scheffner, and P. M. Howley. 1993. Cloning and 16 and 18 promotes the degradation of p53. Cell 63:1129–1136.
expression of the cDNA for E6-AP, a protein that mediates the interaction 54. Schuermann, M., M. Neuberg, J. B. Hunter, T. Jenuwein, R. P. Rysek, R.
of the human papillomavirus E6 oncoprotein with p53. Mol. Cell. Biol. Bravo, and R. Müller. 1989. The leucine repeat motif in Fos protein medi-
13:775–784. ates complex formation with Jun/AP-1 and is required for transformation.
31. Jariel-Encontre, I., M. Pariat, F. Martin, S. Carillo, C. Salvat, and M. Cell 56:507–516.
Piechaczyk. 1995. Ubiquitinylation is not an absolute requirement for deg- 55. Seufert, W., and S. Jentsch. 1990. Ubiquitin-conjugating enzymes UBC4 and
radation of c-Jun by the 26S proteasome. J. Biol. Chem. 270:11623–11627. UBC5 mediate selective degradation of short-lived and abnormal proteins.
32. Jentsch, S. 1992. The ubiquitin conjugating system. Annu. Rev. Genet. EMBO J. 9:543–550.
26:177–205. 56. Shkedy, D., H. Gonen, B. Bercovich, and A. Ciechanover. 1994. Complete
33. Jentsch, S., J. P. McGrath, and A. Varshavsky. 1987. The yeast DNA repair reconstitution of conjugation and subsequent degradation of the tumor sup-
gene RAD6 encodes a ubiquitin-conjugating enzyme. Nature (London) 329: pressor protein p53 by purified components of the ubiquitin proteolytic
131–134. system. FEBS Lett. 348:126–130.
34. Johnson, R. S., B. M. Spiegelman, and V. Papaioannou. 1992. Pleiotropic 57. Traenckner, E. B.-M., S. Wilk, and P. A. Baeuerle. 1994. A proteasome
effects of a null mutation in the c-fos proto-oncogene. Cell 71:577–586. inhibitor prevents activation of NF-kB and stabilizes a newly phosphorylated
35. Johnston, N. L., and R. E. Cohen. 1991. Uncoupling ubiquitin-protein con- form of IkB-a that is still bound to NF-kB. EMBO J. 13:5433–5441.
jugation from ubiquitin-dependent proteolysis by the use of b,g nonhydro- 58. Treier, M., W. Seufert, and S. Jentsch. 1992. Drosophila Ubc1 encodes a
lyzable ATP analogues. Biochemistry 39:7514–7522. highly conserved ubiquitin-conjugating enzyme involved in selective protein
36. Kulka, R. G., B. Raboy, R. Schuster, H. A. Parag, G. Diamond, A. Ciech- degradation. EMBO J. 11:367–372.
anover, and M. Marcus. 1988. A Chinese hamster cell cycle mutant arrested 59. Treier, M., L. M. Staszewski, and D. Bohmann. 1994. Ubiquitin-dependent
at G2 phase has a temperature-sensitive ubiquitin-activating enzyme, E1. J. c-Jun degradation in vivo is mediated by the d domain. Cell 78:787–798.
Biol. Chem. 263:15726–15731. 60. van Beveren, C., F. van Straaten, T. Curran, R. Müller, and I. M. Verma.
37. Maki, Y., T. J. Bos, C. Davis, M. Starbuck, and P. K. Vogt. 1987. Avian 1983. Analysis of FBJ-MuSV provirus and c-fos (mouse) gene reveals that
sarcoma virus 17 carries the jun oncogene. Proc. Natl. Acad. Sci. USA viral and cellular fos gene products have different carboxy termini. Cell
84:2848–2852. 32:1241–1255.
38. Michalek, M. T., E. P. Grant, C. Gramm, A. L. Goldberg, and K. L. Rock. 61. Van Straaten, F., R. Müller, T. Curran, C. van Beveren, and I. M. Verma.
1993. A role for the ubiquitin-dependent proteolytic pathway in MHC class 1983. Complete nucleotide sequence of a human c-onc gene: deduced amino
I-restricted antigen presentation. Nature (London) 363:552–554. acid sequence of the human c-fos gene protein. Proc. Natl. Acad. Sci. USA
39. Murakami, Y., S. Matsufuji, T. Kameji, S.-I. Hayashi, K. Igarashi, T. 80:3183–3187.
Tamura, K. Tanaka, and A. Ichihara. 1992. Ornithine decarboxylase is 62. Wade, W. F., Z. Z. Chen, R. Maki, S. McKercher, E. Palmer, J. C. Cambier,
degraded by the 26S proteasome without ubiquitination. Nature (London) and J. H. Freed. 1989. Altered I-A protein-mediated transmembrane signal-
360:597–599. ing in B cells that express truncated I-Ak protein. Proc. Natl. Acad. Sci. USA
40. Murray, A. 1995. Cyclin ubiquitination: the destructive end of mitosis. Cell 86:6297–6301.
81:149–152. 63. Wang, Z.-Q., C. Ovitt, A. Grigoriadis, U. Mohle-Steinlein, U. Ruther, and
41. Nishizawa, M., K. Okazaki, N. Furuno, N. Watanabe, and N. Segata. 1992. E. F. Wagner. 1992. Bone and hematopoietic defects in mice lacking c-Fos.
The ‘‘second-codon rule’’ and autophosphorylation govern the stability and Nature (London) 360:741–745.
activity of Mos during meiotic cell cycle in Xenopus oocytes. EMBO J. 64. Wong, T. K., and E. Neumann. 1982. Electric field-mediated gene transfer.
11:2433–2446. Biochem. Biophys. Res. Commun. 107:584–587.

You might also like